Skip to main content

Characterization of the human gut virome in metabolic and autoimmune diseases

Abstract

The intestinal microbiome is dominated by bacteria and plays a pivotal role in the occurrence and development of disease, including several metabolic and autoimmune disorders. While intestinal viral communities, primarily made up of bacteriophages, are also thought to play a role in disease pathogenesis in the gastrointestinal tract, they have received much less attention than intestinal bacteria. Thus, there is limited information about the relationship between bacteriophages and disease. This review explores a potential role for the intestinal viral microbiome in various metabolic and autoimmune diseases.

Background

There is a great deal of evidence associating the intestinal bacterial microbiome (bacteriome) with disease pathogenesis. Abnormalities in the human gut microbiota, known as dysbiosis, are linked to a variety of conditions including metabolic disorders, cardiovascular diseases, inflammatory bowel disease (IBD), and autoimmune diseases. Recent studies have shown that trans-kingdom interactions between bacteriomes and viral microbiomes (viromes) are associated with the pathogenesis of pathobiont-mediated diseases [1,2,3,4,5].

It is worth noting that most intestinal viruses are thought to be bacteriophages (phages), which infect and reproduce inside bacteria [6]. Previous studies report that gut phage populations form unique patterns in individuals, independent of genetic factors [6,7,8,9]. While viromes are temporally stable in healthy adults [6,7,8,9], the virome composition changes in diseases such as IBD, which includes Crohn’s disease and ulcerative colitis [10]. Recent work has shown that the intestinal virome develops in infants along with corresponding changes to the bacteriome [11]. In addition, fecal microbiota transplantation (FMT), which is a recommended treatment regimen for Clostridioides difficile infection (CDI) [12], is shown to alter both the bacteriome and the virome [13, 14]. Thus, phages potentially aid bacterial colonization and help to shape bacteriome structure and homeostatic function through host-pathogen interactions [15,16,17,18,19].

There is no universally conserved gene in viruses that is equivalent to the 16S rRNA gene in bacteria or the internal transcribed spacer in fungi, which are used for taxonomic classification [20, 21]. As a result, whole metagenome shotgun sequencing is required to estimate the diversity and taxonomy of intestinal viruses [22], making metagenome research on intestinal phages more challenging than analysis of intestinal bacteria. The vast majority of sequence reads, termed “viral dark matter,” do not align with currently known viral sequences and present a major obstacle to comprehensively defining viromes [23]. This results from a lack of sufficient viral sequence data in public databases because so few intestinal virus analyses have been conducted to date.

Environmental metagenome research, which includes marine virome analysis and intestinal metagenome research, has pioneered technological advances in this field [24,25,26]. Advances in bioinformatics tools for virome analysis, including assemblers such as IDBA-UD, MEGAHIT, and MetaSPAdes [27,28,29], viral sequence detection methods such as VirSorter [18], and contig-based classification tools such as vConTACT [30], have demonstrated that effective contig-based viral classification can be performed [18, 19, 30, 31]. Importantly, multiple displacement amplification (MDA), which has been used for most intestinal virome analyses, was shown to be biased owing to preferential amplification of circular single-stranded DNA (ssDNA), likely explaining why the ssDNA phage, Microviridae, was shown to be dominant in the human intestine [10, 25, 32, 33]. Recently, an Adaptase plus Linker Amplification method was developed to enable the composition of ssDNA viruses to be determined without bias [25]. There was clearly a need to acquire genomic and functional information about the intestinal virome and to further characterize the association between intestinal phages and bacteria using more reliable quantitative methods and informatics tools.

A recent study characterized both the intestinal virome and bacteriome in fecal samples from 101 healthy individuals [3]. By examining the genomic sequences of both viromes and bacteriomes, the bacterial hosts of both temperate and virulent intestinal phages could be effectively determined. Newly identified endolysins associated with Clostridioides difficile were shown to lyse a high-level toxin-producing C. difficile strain and effectively ameliorate acute infection in mice [3]. Thus, comprehensive metagenome analysis can help to understand and prevent microbe-related disorders.

This review summarizes recent advances in the understanding of metabolic disorders, such as metabolic syndrome (MetS), nonalcoholic fatty liver disease (NAFLD), type 2 diabetes mellitus (T2DM), cardiovascular diseases (CVD), and autoimmune diseases, such as type 1 diabetes mellitus (T1DM), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE), with a particular focus on the role of human intestinal viromes.

Role of the intestinal virome in metabolic disorders

While it is well established that the intestinal microbiota contributes to the pathogenesis of metabolic disorders such as obesity, MetS, NAFLD, T2DM, and CVD [34], alterations in the gut virome have also been associated with these diseases. For example, the gut virome was shown to change significantly in mouse models of obesity. The gut viromes in the mucosa of Western diet-fed obese mice are enriched with temperate phages of the Caudovirales order, which associate with the Bacilli, Negativicutes, and Bacteroidia classes. Temperate phages from the Bacteroidia class, in particular, encode stress and niche-specific functions that are advantageous to bacterial host adaptation, supporting the potential of gut phages to impact gut microbial function in response to perturbation [35]. In addition, the viral communities of high-fat diet (HFD)-induced obese mice shifted away from the relatively abundant Siphoviridae towards bacteriophages from the Microviridae family. Particular bacteriophage structural genes declined significantly after the transition to HFD, with a conserved loss of integrase genes, suggesting that these altered viral communities are populated by viruses that are more virulent towards host bacteria [36]. Total viral content in leptin-deficient mice feces is also closely linked with metabolic measures such as body weight, fat mass, and fasting blood glucose and is positively correlated with Firmicutes, a type of bacteria associated with obesity while being negatively correlated with Bacteroides and Bifidobacteria, bacteria associated with leanness [37]. Alterations in the gut virome are thought to have a role in metabolic disorders. Indeed, a study of HFD-induced obese mice showed reduced weight gain and normalized blood glucose tolerance following fecal virome transplant (FVT) from mice with a lean phenotype. FVT significantly changed the bacterial and viral components of the gut microbiota, in addition to the plasma metabolome and expression profiles of obesity and T2DM-associated genes [38]. It was hypothesized that this effect was the result of FVT-induced reshaping of the gut microbiome, warranting further investigation.

Recently, several human studies have sought to characterize the gut virome during various metabolic disorders (Table 1). One study found that obese individuals had lower gut viral richness and diversity and weaker viral-bacterial interactions than lean controls. Eleven viruses, including Escherichia phage, Geobacillus phage, and Lactobacillus phage, were enriched in obese subjects [39]. Another study found higher phage richness and diversity in children with obesity or MetS than in those of normal weight. The phage contigs found at high prevalence in normal-weight individuals were significantly lower in those with obesity and MetS, suggesting that the expansion of specific phages may decrease the concentration of others, and the loss of some phages found at high levels in normal-weight individuals could be associated with obesity and MetS [40]. The dynamics of the gut bacteriophage community after FMT from healthy donors have also been studied in individuals with MetS. Interestingly, the gut virome in patients with better clinical outcomes more closely resembled the viromes of healthy donors [41]. In a study of the gut virome during NAFLD, the liver manifestation of MetS, patients had significantly lower diversity and proportionately fewer phages than control subjects [42]. Lower diversity and bacteriophage number were associated with the severity of NAFLD-associated histologic fibrosis. While the abundance of several Lactococcus and Leuconostoc phages was inversely correlated with liver fibrosis severity, the abundance of Lactobacillus phages correlated positively with fibrosis severity. The abundance of some phages was inversely associated with the abundance of their respective bacterial hosts; however, viral diversity did not always correlate with bacterial diversity, showing that it is difficult to determine what factors are driving lower viral diversity in NAFLD patients.

Table 1 Human studies of the gut virome in metabolic disorders

Ma et al. first identified seven phage operational taxonomic units (pOTUs) (four Siphoviridae, two Podoviridae, and one unclassified family) associated with T2DM patients. However, these T2DM-specific changes in the phageome could not simply be explained as co-variation with their altered bacterial hosts, suggesting that bacteriophages play a complex role in the gut environment that is not limited to interactions with their bacterial hosts [43]. Yang et al. characterized the composition of the gut virome in T2DM patients with obesity and found that these patients had more severe gut viral dysbiosis than those with obesity alone. Although four viral species (Micromonas pusilla virus, Cellulophaga phage, Bacteroides phage, and Halovirus) were higher and 13 viral species (Hokovirus, Klosneuvirus, and Catovirus, etc.) were lower in T2DM subjects with obesity than in lean controls, all these differential viral species had the markers of T2DM, indicating that T2DM was a critical and independent contributor to gut viral dysbiosis during obesity. In addition, obese subjects with T2DM showed a significant decrease in the number of phage-bacterial correlations and a higher negative correlation compared with lean controls, suggesting that strong and complex viral-bacterial trans-kingdom correlations of the gut microbiota in lean controls were lost during obesity and T2DM [39]. An increased abundance of both Enterobacteriaceae-specific phages and Enterobacteriaceae is also reported in the enteric microbiome of T2DM patients, which may increase the amount of circulating lipopolysaccharide (LPS) and the systemic inflammatory response, leading to insulin resistance. In addition, the combination of eight selected phages (three Streptococcus phages, three Enterobacteriaceae-specific phages, one Enterococcus phage, and one Brochothrix phage) reliably differentiated T2DM patients from nondiabetic controls, demonstrating a potential use for the phageome as a diagnostic indicator of T2DM [44].

A limited number of case-control studies have been conducted to characterize the gut virome during CVD. Guo et al. first showed that the percentage of Virgaviridae, a family of rod-shaped plant viruses, was higher, and the number of viruses of the family Microviridae was lower in coronary heart disease (CHD) groups than in control groups, indicating that virome composition may be linked to daily living habits and medical therapy for CHD [45]. A large cohort study of individuals with atherosclerotic CVD (ACVD) also demonstrated that several bacteriophages were more or less abundant in the gut virome of individuals with ACVD than in healthy controls. The known hosts for ACVD-enriched bacteriophages mostly included bacteria from the family Enterobacteriaceae or the genus Streptococcus [46]. In a study of the gut virome in patients with hypertension, a major modifiable risk factor for CVD, two viral types were predominant among the fecal samples, and the primary bacteriophages for viral-type 1 and viral-type 2 were Erwinia phage phiEaH2 and Lactococcus phage 1706, respectively [47]. Additionally, certain viruses, including Cnaphalocrocis medinalis granulovirus, Cronobacter phage CR3, and Streptococcus virus phiAbc2, could be used as biomarkers to distinguish healthy individuals from those with hypertension or prehypertension. The viruses were shown to have a superior resolution and discriminatory power for identifying hypertension than bacteria, suggesting that they may be used for early-stage diagnostics.

Although many metabolic disorders are associated with various gut viruses, it is difficult to establish a consistent direction of change in the gut virome and whether these alterations are the cause or result of each disease. As with bacteria in the gut microbiome, additional large-scale longitudinal trials in humans and translational animal studies, especially using FVT, are required to better assess trends and causality. Future research identifying a role for the gut virome in the etiology of metabolic disorders may help to direct virome-based therapeutic approaches such as novel phage therapy.

Role of the intestinal virome in autoimmune diseases

Recent human studies have investigated the association between the intestinal virome and autoimmune diseases such as T1DM (Table 2). T1DM is characterized by the autoimmune destruction of insulin-producing pancreatic islet beta cells. Most cases of T1DM are preceded by the appearance of islet autoimmunity (IA) that can be serologically confirmed by the presence of at least one autoantibody specific for the pancreatic islets, including insulin (IAA), glutamic acid decarboxylase (GADA), protein tyrosine phosphatase-like insulinoma antigen 2 (IA-2A), and zinc transporter 8 (ZnT8A). While eukaryotic viral infections, such as enterovirus infection, have been proposed as a possible cause of IA and T1DM [54], changes to the gut virome have also been suggested as a potential trigger. The pioneering type 1 diabetes prediction and prevention (DIPP) cohort study of children with IA did not find a significant association between the composition of the gut virome and the development of IA [48]. Indeed, CrAssphage signals showed a possible association between fecal samples from children with early-onset IA followed by T1DM and Bacteroides dorei, which has been associated with the risk of IA [55]. However, no significant correlation between the CrAssphage and IA onset has been identified [49].

Table 2 Human studies of the intestinal virome in autoimmune diseases

Zhao et al. found that two major bacteriophage groups, Myoviridae and Podoviridae, had higher diversity and richness in stool samples from control children than in those from children with IA [50]. Amyloid-producing Escherichia coli (E. coli) in the intestine have also been linked to the development of IA and diabetogenic role of E. coli prophages. An increase in the ratio of E. coli phage to E. coli was observed prior to bacterial host depletion in children who seroconverted or developed T1DM, suggesting that continual induction of prophages or E. coli-harboring prophages may have a fitness advantage in T1DM [51].

While a large number of studies have characterized the gut virome during T1DM, few have characterized the gut virome during systemic autoimmune disease. In a study of individuals at risk for developing rheumatoid arthritis (RA) [52], the composition of the intestinal bacteriophage diverged based on anti-cyclic citrullinated peptide (CCP) autoantibody status, a strong indicator of future RA development. CCP-positive individuals had significantly higher levels of Streptococcaceae, Bacteroidaceae, and Lachnospiraceae phages than healthy controls, indicating the potential of using these phages as biomarkers for preclinical RA. These phages also encoded unique repertoires of auxiliary metabolic genes (AMGs), which modify LPS and other outer membrane glycans on host bacteria, suggesting that they can directly influence the metabolic and immunomodulatory capacity of RA-associated microbiota. Following previous case-control studies of the intestinal bacteriome in Japanese patients with various autoimmune diseases, including RA, systemic lupus erythematosus (SLE), and multiple sclerosis (MS) [56,57,58], Tomofuji et al. recently revealed disease-associated changes in the intestinal virome [53]. In particular, crAss-like phages, which are a primary component of the healthy gut virome, and particular Bacteroidetes and Firmicutes, including Ruminococcus spp., significantly decreased in the gut of patients with RA and SLE. Podoviridae, which has a symbiotic relationship with Faecalibacterium, also declined significantly in the gut of patients with SLE. This evidence suggests that specific phages are associated with autoimmune diseases, possibly by modulating the biological properties of certain bacteria and the host immune system.

Despite great efforts to characterize the gut virome during autoimmune diseases, longitudinal human studies in different populations are needed for a more comprehensive understanding of its role. Novel findings from human studies on the immunomodulatory potential of gut viromes must be confirmed by further investigation using specific T1DM, RA, and SLE animal models.

Conclusions

The intestinal virome has emerged as a pivotal factor responsible for regulating the pathogenesis of intestinal bacteria-mediated diseases. Because many intestinal phages remain uncharacterized, it is difficult to accurately characterize the intestinal virome. However, this will be overcome by further improvements in metagenomic technology, such as the combination of short-read sequencing and long-read sequencing.

Both genetic and environmental factors have a central role in the pathogenesis of metabolic and autoimmune diseases. Therefore, it would be an interesting future direction to elucidate disease mechanisms by integrated analysis of human genomics and intestinal virome.

Since it is necessary to kill only pathobiont without affecting beneficial bacteria in controlling dysbiosis, the host specificity of the bacteriophage deserves attention for this purpose, as in the regulation of multidrug-resistant bacteria. In addition to bacteria, vast numbers of viruses cohabit the human intestine, and a major fraction of the intestinal virome is composed of bacteriophages, whose hosts are commensal bacteria. Therefore, phage therapy is an alternative treatment for the control of dysbiosis and the elimination of pathobionts. Genomic information about intestinal phages is essential for understanding the intestinal bacteria-phage association [3] and will aid the development of next-generation phage therapies. Thus, it is critically important to expand metagenomic technology and its potential clinical applications.

Availability of data and materials

Not applicable

Abbreviations

IBD:

Inflammatory bowel disease

FMT:

Fecal microbiome transplantation

CDI:

Clostridioides difficile infection

MDA:

Multiple displacement amplification

ssDNA:

Single-stranded DNA

MetS:

Metabolic syndrome

NAFLD:

Nonalcoholic fatty liver disease

CVD:

Cardiovascular disease

T1DM:

Type 1 diabetes mellitus

T2DM:

Type 2 diabetes mellitus

RA:

Rheumatoid arthritis

SLE:

Systemic lupus erythematosus

HFD:

High-fat diet

FVT:

Fecal virome transplant

pOTUs:

Phage operational taxonomic units

LPS:

Lipopolysaccharide

CHD:

Coronary heart disease

ACVD:

Atherosclerotic cardiovascular disease

IA:

Islet autoimmunity

IAA:

Insulin autoantibodies

GADA:

Glutamic acid decarboxylase antibodies

IA-2A:

Tyrosine phosphatase-like insulinoma antigen 2

ZnT8A:

Zinc transporter 8 autoantibody

CCP:

Cyclic citrullinated peptide

AMGs:

Auxiliary metabolic genes

MS:

Multiple sclerosis

References

  1. Yutin N, et al. Discovery of an expansive bacteriophage family that includes the most abundant viruses from the human gut. Nat Microbiol. 2018;3(1):38–46.

    Article  CAS  PubMed  Google Scholar 

  2. Shkoporov AN, et al. The human gut virome is highly diverse, stable, and individual specific. Cell Host Microbe. 2019;26(4):527–541 e5.

    Article  CAS  PubMed  Google Scholar 

  3. Fujimoto K, et al. Metagenome data on intestinal phage-bacteria associations aids the development of phage therapy against pathobionts. Cell Host Microbe. 2020;28(3):380–389 e9.

    Article  CAS  PubMed  Google Scholar 

  4. Bajaj JS, et al. Interaction of bacterial metagenome and virome in patients with cirrhosis and hepatic encephalopathy. Gut. 2021;70(6):1162–73.

    Article  CAS  PubMed  Google Scholar 

  5. Fujimoto K, et al. Functional restoration of bacteriomes and viromes by fecal microbiota transplantation. Gastroenterology. 2021;160(6):2089–2102 e12.

    Article  PubMed  Google Scholar 

  6. Manrique P, et al. Healthy human gut phageome. Proc Natl Acad Sci U S A. 2016;113(37):10400–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Minot S, et al. The human gut virome: inter-individual variation and dynamic response to diet. Genome Res. 2011;21(10):1616–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Minot S, et al. Rapid evolution of the human gut virome. Proc Natl Acad Sci U S A. 2013;110(30):12450–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Reyes A, et al. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature. 2010;466(7304):334–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Norman JM, et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell. 2015;160(3):447–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lim ES, et al. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015;21(10):1228–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. McDonald LC, et al. Clinical practice guidelines for Clostridium difficile Infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis. 2018;66(7):987–94.

    Article  CAS  PubMed  Google Scholar 

  13. Kang DW, et al. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5(1):10.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zuo T, et al. Bacteriophage transfer during faecal microbiota transplantation in Clostridium difficile infection is associated with treatment outcome. Gut. 2018;67(4):634–43.

    CAS  PubMed  Google Scholar 

  15. De Paepe M, et al. Bacteriophages: an underestimated role in human and animal health? Front Cell Infect Microbiol. 2014;4:39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Edwards RA, et al. Computational approaches to predict bacteriophage-host relationships. FEMS Microbiol Rev. 2016;40(2):258–72.

    Article  CAS  PubMed  Google Scholar 

  17. Mills S, et al. Movers and shakers: influence of bacteriophages in shaping the mammalian gut microbiota. Gut Microbes. 2013;4(1):4–16.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Roux S, et al. Viral dark matter and virus-host interactions resolved from publicly available microbial genomes. Elife. 2015;4:e08490.

  19. Roux S, et al. Ecogenomics and potential biogeochemical impacts of globally abundant ocean viruses. Nature. 2016;537(7622):689–93.

    Article  CAS  PubMed  Google Scholar 

  20. Schoch CL, et al. Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for fungi. Proc Natl Acad Sci U S A. 2012;109(16):6241–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stackebrandt E, Goebel BM. A place for DNA-DNA reassociation and 16s ribosomal-RNA sequence-analysis in the present species definition in bacteriology. Int J Syst Bacteriol. 1994;44(4):846–9.

    Article  CAS  Google Scholar 

  22. Thurber RV, et al. Laboratory procedures to generate viral metagenomes. Nat Protoc. 2009;4(4):470–83.

    Article  CAS  PubMed  Google Scholar 

  23. Hurwitz BL, U'Ren JM, Youens-Clark K. Computational prospecting the great viral unknown. FEMS Microbiol Lett. 2016;363(10):fnw077.

  24. Duhaime MB, et al. Towards quantitative metagenomics of wild viruses and other ultra-low concentration DNA samples: a rigorous assessment and optimization of the linker amplification method. Environ Microbiol. 2012;14(9):2526–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Roux S, et al. Towards quantitative viromics for both double-stranded and single-stranded DNA viruses. PeerJ. 2016;4:e2777.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Solonenko SA, et al. Sequencing platform and library preparation choices impact viral metagenomes. BMC Genomics. 2013;14:320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li D, et al. MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics. 2015;31(10):1674–6.

    Article  CAS  PubMed  Google Scholar 

  28. Nurk S, et al. metaSPAdes: a new versatile metagenomic assembler. Genome Res. 2017;27(5):824–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Peng Y, et al. IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. Bioinformatics. 2012;28(11):1420–8.

    Article  CAS  PubMed  Google Scholar 

  30. Bolduc B, et al. vConTACT: an iVirus tool to classify double-stranded DNA viruses that infect archaea and bacteria. PeerJ. 2017;5:e3243.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Bolduc B, et al. iVirus: facilitating new insights in viral ecology with software and community data sets imbedded in a cyberinfrastructure. ISME J. 2017;11(1):7–14.

    Article  PubMed  Google Scholar 

  32. Binga EK, Lasken RS, Neufeld JD. Something from (almost) nothing: the impact of multiple displacement amplification on microbial ecology. ISME J. 2008;2(3):233–41.

    Article  CAS  PubMed  Google Scholar 

  33. Marine R, et al. Caught in the middle with multiple displacement amplification: the myth of pooling for avoiding multiple displacement amplification bias in a metagenome. Microbiome. 2014;2(1):3.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71.

    Article  CAS  PubMed  Google Scholar 

  35. Kim MS, Bae JW. Spatial disturbances in altered mucosal and luminal gut viromes of diet-induced obese mice. Environ Microbiol. 2016;18(5):1498–510.

    Article  CAS  PubMed  Google Scholar 

  36. Schulfer A, et al. Fecal viral community responses to high-fat diet in mice. mSphere. 2020;5(1):e00833–19 .

  37. Yadav H, et al. Increased fecal viral content associated with obesity in mice. World J Diabetes. 2016;7(15):316–20.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Rasmussen TS, et al. Faecal virome transplantation decreases symptoms of type 2 diabetes and obesity in a murine model. Gut. 2020;69(12):2122–30.

    Article  CAS  PubMed  Google Scholar 

  39. Yang K, et al. Alterations in the gut virome in obesity and type 2 diabetes mellitus. Gastroenterology. 2021;161(4):1257–1269 e13.

    Article  CAS  PubMed  Google Scholar 

  40. Bikel S, et al. Gut dsDNA virome shows diversity and richness alterations associated with childhood obesity and metabolic syndrome. iScience. 2021;24(8):102900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Manrique P, et al. Gut bacteriophage dynamics during fecal microbial transplantation in subjects with metabolic syndrome. Gut Microbes. 2021;13(1):1–15.

    Article  PubMed  CAS  Google Scholar 

  42. Lang S, et al. Intestinal virome signature associated with severity of nonalcoholic fatty liver disease. Gastroenterology. 2020;159(5):1839–52.

    Article  CAS  PubMed  Google Scholar 

  43. Ma Y, et al. A human gut phage catalog correlates the gut phageome with type 2 diabetes. Microbiome. 2018;6(1):24.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Chen Q, et al. Enteric phageome alterations in patients with type 2 diabetes. Front Cell Infect Microbiol. 2020;10:575084.

    Article  PubMed  Google Scholar 

  45. Guo L, et al. Viral metagenomics analysis of feces from coronary heart disease patients reveals the genetic diversity of the Microviridae. Virol Sin. 2017;32(2):130–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jie Z, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 2017;8(1):845.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Han M, et al. The human gut virome in hypertension. Front Microbiol. 2018;9:3150.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Kramna L, et al. Gut virome sequencing in children with early islet autoimmunity. Diabetes Care. 2015;38(5):930–3.

    Article  CAS  PubMed  Google Scholar 

  49. Cinek O, et al. Imbalance of bacteriome profiles within the Finnish diabetes prediction and prevention study: parallel use of 16S profiling and virome sequencing in stool samples from children with islet autoimmunity and matched controls. Pediatr Diabetes. 2017;18(7):588–98.

    Article  CAS  PubMed  Google Scholar 

  50. Zhao G, et al. Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children. Proc Natl Acad Sci U S A. 2017;114(30):E6166–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tetz G, et al. Type 1 diabetes: an association between autoimmunity, the dynamics of gut amyloid-producing E. coli and their phages. Sci Rep. 2019;9(1):9685.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Mangalea MR, et al. Individuals at risk for rheumatoid arthritis harbor differential intestinal bacteriophage communities with distinct metabolic potential. Cell Host Microbe. 2021;29(5):726–739 e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tomofuji Y, et al. Whole gut virome analysis of 476 Japanese revealed a link between phage and autoimmune disease. Ann Rheum Dis. 2022;81(2):278–88.

    Article  PubMed  Google Scholar 

  54. Yeung WC, Rawlinson WD, Craig ME. Enterovirus infection and type 1 diabetes mellitus: systematic review and meta-analysis of observational molecular studies. BMJ. 2011;342:d35.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Davis-Richardson AG, et al. Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front Microbiol. 2014;5:678.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kishikawa T, et al. Metagenome-wide association study of gut microbiome revealed novel aetiology of rheumatoid arthritis in the Japanese population. Ann Rheum Dis. 2020;79(1):103–11.

    Article  CAS  PubMed  Google Scholar 

  57. Tomofuji Y, et al. Metagenome-wide association study revealed disease-specific landscape of the gut microbiome of systemic lupus erythematosus in Japanese. Ann Rheum Dis. 2021;80(12):1575–83.

    Article  CAS  PubMed  Google Scholar 

  58. Kishikawa T, et al. A metagenome-wide association study of gut microbiome in patients with multiple sclerosis revealed novel disease pathology. Front Cell Infect Microbiol. 2020;10:585973.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Ogawa, M. Maeda, and K. Suetsugu for secretarial assistance. We thank the Edanz (https://jp.edanz.com/ac) for editing a draft of this manuscript.

Funding

This study was supported by the Japan Agency for Medical Research and Development (AMED) (to K. F., 21ae0121048h0001, to S. U., 21fk0108619h0001).

Author information

Authors and Affiliations

Authors

Contributions

KF and DM wrote the draft and created the table. SU supervised the study and drafted the manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Satoshi Uematsu.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fujimoto, K., Miyaoka, D. & Uematsu, S. Characterization of the human gut virome in metabolic and autoimmune diseases. Inflamm Regener 42, 32 (2022). https://doi.org/10.1186/s41232-022-00218-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41232-022-00218-6

Keywords